Next Article in Journal
Practical School Nutrition Program May Reduce Food Neophobia
Previous Article in Journal
Plant-Based Dietary Practices and Socioeconomic Factors That Influence Anemia in India
Previous Article in Special Issue
Low Circulating Concentrations of Very Long Chain Saturated Fatty Acids Are Associated with High Risk of Mortality in Kidney Transplant Recipients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Fatty Acids and Frailty: A Mendelian Randomization Study

1
Department of Medical Epidemiology and Biostatistics, Karolinska Institute, 171 77 Stockholm, Sweden
2
Faculty of Health and Social Services, School of Nutrition and Dietetics, Kanagawa University of Human Services, Yokosuka 238-8522, Japan
3
Gerontology Research Center (GEREC), Faculty of Social Sciences (Health Sciences), University of Tampere, 33014 Tampere, Finland
*
Author to whom correspondence should be addressed.
Nutrients 2021, 13(10), 3539; https://doi.org/10.3390/nu13103539
Submission received: 31 August 2021 / Revised: 1 October 2021 / Accepted: 6 October 2021 / Published: 9 October 2021
(This article belongs to the Special Issue Epidemiological Study of Fatty Acids for Healthy Ageing)

Abstract

:
Background: Observational studies have suggested that fatty acids such as higher levels of n-3 polyunsaturated fatty acids (PUFAs) may prevent frailty. By using Mendelian randomization analysis, we examined the relationship between fatty acids and frailty. Methods: We used summary statistics data for single-nucleotide polymorphisms associated with plasma levels of saturated fatty acids (palmitic acid, stearic acid), mono-unsaturated fatty acids (MUFAs) (palmitoleic acid, oleic acid), n-6 PUFAs (linoleic acid, arachidonic acid), and n-3 PUFAs (alpha-linolenic acid, eicosapentaenoic acid, docosapentaenoic acid, docosahexaenoic acid), and the corresponding data for frailty index (FI) in 356,432 individuals in the UK Biobank. Results: Although there were no robust associations on the MUFAs or the PUFAs, genetically predicted higher plasma stearic acid level (one of saturated fatty acids) was statistically significantly associated with higher FI (β = 0.178; 95% confidence interval = −0.050 to 0.307; p = 0.007). Such a relationship was also observed in a multivariate MR (β = 0.361; 95% confidence interval = 0.155 to 0.567; p = 0.001). Genetically predicted higher palmitic acid was also significantly associated with higher FI (β = 0.288; 95% confidence interval = 0.128 to 0.447; p < 0.001) in the multivariate MR analysis. Conclusions: The present MR study implies that saturated fatty acids, especially stearic acid, is a risk factor of frailty.

1. Introduction

Fatty acids are one of the nutritional components that contribute to healthy aging [1]. For example, n-3 polyunsaturated fatty acids (PUFAs) such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) are known to reduce inflammation, suggesting a beneficial role in various therapeutic areas [2]. There is, however, sufficient evidence for an anti-inflammatory effect on aging only for n-3 PUFA intake [3]. As for n-3 PUFAs, evidence on benefits for cognitive health and age- and disease-related decline in muscle mass has been presented [2,4,5,6].
There is some evidence from observational studies showing an association between fatty acids and frailty [7]. The overall results suggest that n-3 PUFAs are inversely associated with the risk of frailty [8]. The observational study using U.S. nationally representative data (the National Health and Nutrition Examination Survey) also showed that low n-3 PUFA intake (DHA and EPA) was associated with a higher value of frailty index (FI) in participants aged ≥20 years. In addition, mono-unsaturated fatty acids (MUFAs) were also suggested as preventive factors for frailty [9]. On the other hand, higher saturated fatty acid intake was associated with higher risk of both frailty and mortality even after considering the degree of nutritional deficits [10]. Thus, in conclusion, various types of fatty acids may contribute to frailty.
However, observational studies may have methodological problems, such as confounding or reverse causality. For example, a pooled analysis with data from cohort studies showed that higher n-3 PUFA levels were significantly associated with a lower risk of cardiovascular disease-related deaths [11]. However, a meta-analysis of randomized controlled trials showed that n-3 PUFA had no significant association with cardiovascular disease [12]. The discrepancies in these results may be explained by residual confounding or reverse causality in observational studies [11]. An RCT is more suitable to examine causal relationships without confounding concerns [12]. However, regarding frailty, no randomized controlled trial (RCT) has yet been performed to analyze the effect of fatty acids.
To overcome the problem of confounding and reverse causality in observational studies, Mendelian randomization (MR) is becoming widespread for assessing whether the association is consistent with a causal hypothesis. An MR study is a type of instrumental variable analysis where genetic variants (single-nucleotide polymorphisms; SNPs) are applied as instrumental variables for the exposure. An MR study is often described as a “natural RCT” because random allocation of alleles during meiosis is conceptually similar to RCT design. Therefore, an MR study would provide more robust evidence regarding the causal relationship between fatty acids and frailty than observational studies. However, to our knowledge, no MR study has yet investigated the associations between fatty acids and frailty, although there have been several MR studies on the relationship between fatty acids and cardiovascular disease [13,14,15,16,17].
The aim of the present MR study was to examine the associations between fatty acids and frailty.

2. Materials and Methods

2.1. Study Design

Two-sample MR analyses using summary statistics of a genome-wide association study (GWAS) were conducted. As exposure measures, we examined ten types of plasma fatty acids: two types of saturated fatty acids (palmitic acid, stearic acid), two types of MUFAs (palmitoleic acid, oleic acid), two types of n-6 PUFAs (linoleic acid, arachidonic acid), and four types of n-3 PUFAs (α-linolenic acid, eicosapentaenoic acid, docosapentaenoic acid, docosahexaenoic acid) [18]. As an outcome measure of frailty, the Rockwood FI was used [19].

2.2. Data Sources

For estimating the genotype–FI associations, we analyzed individual-level data from participants enrolled in the UK Biobank [20]. The UK Biobank is a population-based cohort study on individuals aged from 40 to 69 years recruited from the UK National Health Service registers. The UK Biobank includes data on genetic and environmental factors across the UK collected from 2006 to 2010 (https://www.ukbiobank.ac.uk/ [accecced on 3 March 2021]). Among the 500,336 participants who had information about FI available [19], we included 356,432 participants who were (1) genotyped, (2) of European ancestry, (3) had complete information on variables included in the FI, and (4) not randomly dropped with a kinship threshold of 0.1768 for considering relatedness. The analytical sample comprised 189,949 women (53.3%) and 166,483 men (46.7%), with a mean (SD) age of 56.7 (8.0) years.
We used summary statistics data from the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) consortium to examine the association between genetic variants and plasma levels of fatty acids [21,22,23]. The summary statistics were based on a meta-analysis of GWAS of individuals of European ancestry (n  =  8961 individuals for saturated fatty acids or mono-unsaturated fatty acids, n  =  8631 individuals for n-6 PUFAs, and n  =  8866 individuals for n-3 PUFAs). Age ranged from 21 to 102 years [23].

2.3. Selection of Instrumental Variables

We selected SNPs associated with the plasma fatty acids (p < 5 × 10−8) in the previous GWAS meta-analysis in individuals of European ancestry (Table 1) [21,22,23]. As one SNP (rs11190604) was not available in the UK Biobank data, we used an overlapping proxy SNP (rs7099965) in high-linkage disequilibrium (r2 = 1) identified using the LDproxy search on the online platform LDlink (https://ldlink.nci.nih.gov/ [accecced on 3 March 2021]). These SNPs have been used as instrumental variables for plasma fatty acids in previous MR studies [18] and were not considered as weak instrumental variables (F statistic ≥ 10) [24].
To consider the possibility that palindromic SNPs alter the results associated with frailty in several fatty acids (stearic acid, linoleic acid, eicosapentaenoic acid), we also used proxy SNPs in high-linkage disequilibrium (r2 > 0.90) instead of three SNPs; rs11119805, rs10740118, and rs3798713 (Table 2).

2.4. Frailty

The Rockwood FI, based on the accumulation of deficits model, was used as an outcome measure of frailty. The FI is a continuous measure with a high sensitivity also at the lower end of the frailty continuum. According to the principles of this model [25], the FI value for each individual was calculated as the number of deficits present divided by the total number of 49 deficits as described in a previous study [19]. In the present study, the FI value was expressed as a percentage (%). For example, an individual having 10 deficits has an FI of 10/49 = 0.204 (20.4%).

2.5. Statistical Analysis

We estimated the association of each genetic variant with the continuous FI using linear regression analyses to obtain summary statistics for the two-sample MR analyses. All the regression analyses were adjusted for age (continuous variable) and sex using SAS version 9.4 (SAS Inc., Cary, NC, USA).
Two-sample MR analyses were performed to calculate the coefficients (linear association) and 95% confidence intervals (CIs) for FI. All MR analyses were performed by using the “MendelianRandomization” package for R version 3.4.3 (The R Foundation for Statistical Computing). We primarily used the inverse variance weighted method with fixed-effect standard errors [26]. The inverse variance weighted method assumes that all variants are valid instrumental variables.
In addition, we also conducted sensitivity analyses using the MR-Egger method for exposures that used three or more SNPs as instrumental variables (i.e., stearic acid, palmitoleic acid, linoleic acid, docosapentaenoic acid). The MR-Egger method estimates the effect size by adjusting for horizontal pleiotropy (i.e., the genetic variants have effects on the outcome through other paths than via the exposure of interest) [26]. Pleiotropy was assessed using the MR-Egger intercept test, which assumes that the intercept should be zero if the genotype–exposure association is zero.

3. Results

3.1. Summary Statistics

By using the linear regression analyses, we obtained summary statistics for the associations between the genetic variants and FI that were needed to conduct the MR analysis (Table 1 and Table 2).

3.2. Main Results

The results of the MR analyses based on the inverse variance weighted method are shown in Table 3.
In the non-PUFAs, genetically predicted plasma stearic acid level was statistically significantly associated with higher value of FI in both the MR (β = 0.178; 95%CI = −0.050 to 0.307; p = 0.007) and the multivariate MR (β = 0.361; 95%CI = 0.155 to 0.567; p = 0.001). In the multivariate MR analysis, palmitic acid was also significantly associated with higher FI (β = 0.288; 95%CI = 0.128 to 0.447; p < 0.001).
Of the PUFAs, none of the genetically predicted plasma PUFA levels were statistically significantly associated with FI in the multivariate MR.
There was no evidence for pleiotropy based on the MR-Egger analyses in all four types of fatty acids—stearic acid, palmitoleic acid, linoleic acid, and docosapentaenoic acid (P-intercept > 0.05).

3.3. Sensitivity Analysis

The results of the sensitivity MR analyses using proxy SNPs are shown in Table 4. Genetically predicted plasma stearic acid level was statistically significantly associated with higher FI in both the MR (β = 0.177; 95%CI = 0.049 to 0.305; p = 0.007) and the multivariate MR (β = 0.360; 95%CI = 0.154 to 0.566; p = 0.001).

4. Discussion

The present study is an MR study that examines the relationship between genetically predicted levels of fatty acids and frailty. Our results showed that two major saturated fatty acids (palmitic acid, stearic acid) were statistically significantly associated with higher FI even after using the multivariate MR. However, there were no robust associations on the major types of MUFAs or PUFAs (e.g., oleic acid, eicosapentaenoic acid, docosahexaenoic acid) in the multivariate MR.
In the results of the present study, higher stearic acid was consistently associated with higher FI in both the MR and the multivariate MR (Table 3). Furthermore, the MR-Egger analysis suggested no evidence that the result of stearic acid-induced pleiotropy. The results of the multivariate MR showed that palmitic acid was also associated with a higher FI. Thus, our results support the hypothesis that higher levels of saturated fatty acids are risk factors for frailty. Stearic acid and palmitic acid are the major dietary saturated fatty acids typically reflected by adherence to a Western diet including a relatively high amount of animal fat [27]. A previous observational study reported that a higher percentage of saturated fatty acid intake was associated with a higher risk of both frailty and mortality, even after considering the degree of nutritional deficits [10]. A population-based cohort study suggested that the animal protein intake was significantly associated with a higher FI in adults (aged ≥55 years) of normal body weight, and saturated fatty acids tended to be associated with a higher FI in the same population [28]. The observational study revealed that a Westernized-type dietary pattern (characterized by the highest saturated fats) was strongly associated with a higher frailty prevalence, whereas Mediterranean-type dietary pattern (characterized by relatively low intake of meat, egg, fish, and dairy products) was inversely associated with the prevalence of frailty [29]. Another cohort study also revealed that a Westernized dietary pattern (characterized by a high intake of refined bread, whole dairy products, and red and processed meat, as well as low consumption of fruit and vegetables) tended to be associated with a higher risk of frailty [30]. Meta-analyses of observational studies showed associations between saturated fatty acids intake and a high risk of coronary heart disease mortality [31], bone fracture [32], or cognitive disorders [33]. Indeed, a MR study based on UK biobank data revealed that of the major fatty acids, stearic acid was associated with a high risk of cardiovascular diseases such as aortic valve stenosis and venous thrombosis [13]. Another MR study based on UK biobank data also revealed that stearic acid was significantly associated with lower bone mineral density and a higher risk of fracture [34]. Furthermore, MR study-based DIAGRAM consortium including UK biobank data showed that stearic acid was significantly associated with a higher risk of type 2 diabetes. Hence, it would be expected that reduction in the dietary intake of saturated fatty acids contributes to the prevention of frailty via multiple traits. However, based on the current evidence, it is unclear whether dietary saturated fatty acids should be substituted by MUFAs or PUFAs. For example, a previous cohort study reported that substitution of saturated fatty acids (palmitic acid and stearic acid) with plant proteins resulted in a reduction of myocardial infarction risk, but not substitution with MUFAs or PUFAs [35].
However, it is open for discussion whether stearic acid is a specific risk factor of most frailty components or not. For example, a previous cohort study reported that plasma stearic acid was associated with a higher risk of colorectal cancer, although dietary palmitic acid was inversely associated with colorectal cancer [36]. A previous MR study also supported that stearic acid (but not palmitic acid) increases colorectal cancer risk [18]. However, one previous cohort study reported that higher concentrations of plasma phospholipid stearic acid were associated with a lower risk of all-cause mortality, although palmitic acid was associated with a higher risk of total mortality [37]. For example, as for cardiometabolic risk factors, substituting palmitic acid with stearic acid lowers LDL-cholesterol concentrations [38]. Thus, further studies are needed to verify whether only stearic acid is a risk factor among the saturated fatty acids.
As one of major methodological issues about bias towards the null in MR studies, weak instrument bias is often suggested. Although we used only 1 to 3 SNPs as instrumental variables for each PUFA, the SNPs fulfilled the criterion as not being weak instrumental variables (F statistic > 10). However, the change in coefficients and 95%CIs between the MR and the multivariate MR (Table 3) may not be interpreted as robust estimates.
Our study had several strengths. First, to our knowledge, the present study is the first MR study to examine the associations between fatty acids and frailty. This approach (MR study) is less prone to confounding compared to conventional observational studies. Second, we conducted several sensitivity analyses to check robustness of the main results such as the multivariate MR. The present study has also several limitations. First, we only used 1 to 3 SNPs as instrumental variables for each PUFA. Therefore, only four types of fatty acids (stearic acid, palmitoleic acid, linoleic acid, docosapentaenoic acid) could be used for pleiotropy test by the MR-Egger intercept test. Second, because shared SNPs were used among the fatty acids (e.g., rs174547 was used for LA, AA, ALA, and DPA), identifying the effects for individual fatty acids was difficult. Although we thereby conducted the multivariate MR to overcome this problem, these results would not be robust estimates as stated before. Third, because our analysis was restricted to populations of European ancestry, the present findings may not apply to non-European populations.

5. Conclusions

The present MR study suggested that saturated fatty acids (especially stearic acid) is a risk factor of frailty. Nevertheless, future studies may need to consider the balance of fatty acid composition rather than the effect of a single component of fatty acid.

Author Contributions

Conceptualization, Y.T., S.H. and J.J.; methodology, Y.T., S.H. and J.J.; software, Y.T. and Y.W.; formal analysis, Y.T.; resources, Y.W. and J.J.; data curation, J.J.; writing—original draft preparation, Y.T.; writing—review and editing, Y.W., S.H. and J.J.; visualization, Y.T.; supervision, J.J.; project administration, J.J.; funding acquisition, S.H. and J.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Swedish Research Council (grant number 2018-02077, 2019-01272), the Loo & Hans Osterman Foundation, the Strategic Research Program in Epidemiology at Karolinska Institutet, and King Gustaf V’s and Queen Victoria’s Freemason Foundation.

Institutional Review Board Statement

Ethical approval for this study is covered by the general ethics review for UKB, conducted by the North West Haydock Research Ethics Committee of the UK’s Health Research Authority (Reference 16/NW/0274, 13 May 2016). In addition, a permit was approved by the Swedish Ethical Review Authority in Stockholm for conducting these studies in Sweden (Dnr. 2016/1888-31/1).

Informed Consent Statement

Informed consent was obtained from all subjects of UK Biobank involved in the study.

Data Availability Statement

This research has been conducted using the UK Biobank Resource under Application Number 22224. The analyses on UK Biobank genotypes were enabled by resources provided by the Swedish National Infrastructure for Computing (SNIC) at Uppmax partially funded by the Swedish Research Council through grant agreement no. 2018-05973. The data underlying the results presented in the study are available from the UK Biobank (http://biobank.ndph.ox.ac.uk/showcase/ [accecced on 1 October 2021]).

Acknowledgments

We thank the UK Biobank participants, the UK Biobank for making the data available.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ruxton, C.H.; Derbyshire, E.; Toribio-Mateas, M. Role of fatty acids and micronutrients in healthy ageing: A systematic review of randomised controlled trials set in the context of European dietary surveys of older adults. J. Hum. Nutr. Diet. 2016, 29, 308–324. [Google Scholar] [CrossRef]
  2. Troesch, B.; Eggersdorfer, M.; Laviano, A.; Rolland, Y.; Smith, A.D.; Warnke, I.; Weimann, A.; Calder, P.C. Expert Opinion on Benefits of Long-Chain Omega-3 Fatty Acids (DHA and EPA) in Aging and Clinical Nutrition. Nutrients 2020, 12, 2555. [Google Scholar] [CrossRef]
  3. Ticinesi, A.; Meschi, T.; Lauretani, F.; Felis, G.; Franchi, F.; Pedrolli, C.; Barichella, M.; Benati, G.; Di Nuzzo, S.; Ceda, G.P.; et al. Nutrition and Inflammation in Older Individuals: Focus on Vitamin D, n-3 Polyunsaturated Fatty Acids and Whey Proteins. Nutrients 2016, 8, 186. [Google Scholar] [CrossRef] [Green Version]
  4. Yurko-Mauro, K.; Alexander, D.D.; Van Elswyk, M.E. Docosahexaenoic acid and adult memory: A systematic review and meta-analysis. PLoS ONE 2015, 10, e0120391. [Google Scholar] [CrossRef] [Green Version]
  5. Huang, Y.H.; Chiu, W.C.; Hsu, Y.P.; Lo, Y.L.; Wang, Y.H. Effects of Omega-3 Fatty Acids on Muscle Mass, Muscle Strength and Muscle Performance among the Elderly: A Meta-Analysis. Nutrients 2020, 12, 3739. [Google Scholar] [CrossRef] [PubMed]
  6. Strike, S.C.; Carlisle, A.; Gibson, E.L.; Dyall, S.C. A High Omega-3 Fatty Acid Multinutrient Supplement Benefits Cognition and Mobility in Older Women: A Randomized, Double-blind, Placebo-controlled Pilot Study. J. Gerontol. A: Biol. Sci. Med. Sci. 2016, 71, 236–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Hernández Morante, J.J.; Gómez Martínez, C.; Morillas-Ruiz, J.M. Dietary Factors Associated with Frailty in Old Adults: A Review of Nutritional Interventions to Prevent Frailty Development. Nutrients 2019, 11, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Kim, D.; Won, C.W.; Park, Y. Association Between Erythrocyte Levels of n-3 Polyunsaturated Fatty Acids and Risk of Frailty in Community-Dwelling Older Adults: The Korean Frailty and Aging Cohort Study. J. Gerontol. A: Biol. Sci. Med. Sci. 2021, 76, 499–504. [Google Scholar] [CrossRef] [PubMed]
  9. Sandoval-Insausti, H.; Pérez-Tasigchana, R.F.; López-García, E.; García-Esquinas, E.; Rodríguez-Artalejo, F.; Guallar-Castillón, P. Macronutrients Intake and Incident Frailty in Older Adults: A Prospective Cohort Study. J. Gerontol. A: Biol. Sci. Med. Sci. 2016, 71, 1329–1334. [Google Scholar] [CrossRef] [PubMed]
  10. Jayanama, K.; Theou, O.; Godin, J.; Cahill, L.; Rockwood, K. Association of fatty acid consumption with frailty and mortality among middle-aged and older adults. Nutrition 2020, 70, 110610. [Google Scholar] [CrossRef] [PubMed]
  11. Harris, W.S.; Tintle, N.L.; Imamura, F.; Qian, F.; Korat, A.V.A.; Marklund, M.; Djoussé, L.; Bassett, J.K.; Carmichael, P.H.; Chen, Y.Y.; et al. Blood n-3 fatty acid levels and total and cause-specific mortality from 17 prospective studies. Nat. Commun. 2021, 12, 2329. [Google Scholar] [CrossRef]
  12. Aung, T.; Halsey, J.; Kromhout, D.; Gerstein, H.C.; Marchioli, R.; Tavazzi, L.; Geleijnse, J.M.; Rauch, B.; Ness, A.; Galan, P.; et al. Associations of Omega-3 Fatty Acid Supplement Use With Cardiovascular Disease Risks: Meta-analysis of 10 Trials Involving 77 917 Individuals. JAMA Cardiol. 2018, 3, 225–234. [Google Scholar] [CrossRef] [Green Version]
  13. Yuan, S.; Bäck, M.; Bruzelius, M.; Mason, A.M.; Burgess, S.; Larsson, S. Plasma Phospholipid Fatty Acids, FADS1 and Risk of 15 Cardiovascular Diseases: A Mendelian Randomisation Study. Nutrients 2019, 11, 3001. [Google Scholar] [CrossRef] [Green Version]
  14. Yuan, T.; Si, S.; Li, Y.; Li, W.; Chen, X.; Liu, C.; Li, J.; Wang, B.; Hou, L.; Liu, Y.; et al. Roles for circulating polyunsaturated fatty acids in ischemic stroke and modifiable factors: A Mendelian randomization study. Nutr. J. 2020, 19, 70. [Google Scholar] [CrossRef]
  15. Park, S.; Lee, S.; Kim, Y.; Lee, Y.; Kang, M.; Kim, K.; Kim, Y.; Han, S.; Lee, H.; Lee, J.; et al. Causal Effects of Serum Levels of n-3 or n-6 Polyunsaturated Fatty Acids on Coronary Artery Disease: Mendelian Randomization Study. Nutrients 2021, 13, 1490. [Google Scholar] [CrossRef]
  16. Ma, M.; Yang, F.; Wang, Z.; Bao, Q.; Shen, J.; Xie, X. Association of plasma polyunsaturated fatty acids with arterial blood pressure: A Mendelian randomization study. Medicine 2021, 100, e24359. [Google Scholar] [CrossRef]
  17. Zhang, T.; Zhao, J.V.; Schooling, C.M. The associations of plasma phospholipid arachidonic acid with cardiovascular diseases: A Mendelian randomization study. EBioMedicine 2021, 63, 103189. [Google Scholar] [CrossRef] [PubMed]
  18. May-Wilson, S.; Sud, A.; Law, P.J.; Palin, K.; Tuupanen, S.; Gylfe, A.; Hanninen, U.A.; Cajuso, T.; Tanskanen, T.; Kondelin, J.; et al. Pro-inflammatory fatty acid profile and colorectal cancer risk: A Mendelian randomisation analysis. Eur. J. Cancer 2017, 84, 228–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Williams, D.M.; Jylhävä, J.; Pedersen, N.L.; Hägg, S. A Frailty Index for UK Biobank Participants. J. Gerontol. A: Biol. Sci. Med. Sci. 2019, 74, 582–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Sudlow, C.; Gallacher, J.; Allen, N.; Beral, V.; Burton, P.; Danesh, J.; Downey, P.; Elliott, P.; Green, J.; Landray, M.; et al. UK biobank: An open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 2015, 12, e1001779. [Google Scholar] [CrossRef] [Green Version]
  21. Wu, J.H.; Lemaitre, R.N.; Manichaikul, A.; Guan, W.; Tanaka, T.; Foy, M.; Kabagambe, E.K.; Djousse, L.; Siscovick, D.; Fretts, A.M.; et al. Genome-wide association study identifies novel loci associated with concentrations of four plasma phospholipid fatty acids in the de novo lipogenesis pathway: Results from the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) consortium. Circ. Cardiovasc. Genet. 2013, 6, 171–183. [Google Scholar] [CrossRef] [Green Version]
  22. Guan, W.; Steffen, B.T.; Lemaitre, R.N.; Wu, J.H.Y.; Tanaka, T.; Manichaikul, A.; Foy, M.; Rich, S.S.; Wang, L.; Nettleton, J.A.; et al. Genome-wide association study of plasma N6 polyunsaturated fatty acids within the cohorts for heart and aging research in genomic epidemiology consortium. Circ. Cardiovasc. Genet. 2014, 7, 321–331. [Google Scholar] [CrossRef] [Green Version]
  23. Lemaitre, R.N.; Tanaka, T.; Tang, W.; Manichaikul, A.; Foy, M.; Kabagambe, E.K.; Nettleton, J.A.; King, I.B.; Weng, L.C.; Bhattacharya, S.; et al. Genetic loci associated with plasma phospholipid n-3 fatty acids: A meta-analysis of genome-wide association studies from the CHARGE Consortium. PLoS Genet. 2011, 7, e1002193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Liyanage, U.E.; Law, M.H.; Ong, J.S.; Cust, A.E.; Mann, G.J.; Ward, S.V.; Gharahkhani, P.; Iles, M.M.; MacGregor, S. Polyunsaturated fatty acids and risk of melanoma: A Mendelian randomisation analysis. Int. J. Cancer 2018, 143, 508–514. [Google Scholar] [CrossRef] [PubMed]
  25. Searle, S.D.; Mitnitski, A.; Gahbauer, E.A.; Gill, T.M.; Rockwood, K. A standard procedure for creating a frailty index. BMC Geriatr. 2008, 8, 24. [Google Scholar] [CrossRef] [Green Version]
  26. Bowden, J.; Davey Smith, G.; Burgess, S. Mendelian randomization with invalid instruments: Effect estimation and bias detection through Egger regression. Int. J. Epidemiol. 2015, 44, 512–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Iggman, D.; Risérus, U. Role of different dietary saturated fatty acids for cardiometabolic risk. Clin. Lipidol. 2011, 6, 209–223. [Google Scholar] [CrossRef]
  28. Verspoor, E.; Voortman, T.; van Rooij, F.J.A.; Rivadeneira, F.; Franco, O.H.; Kiefte-de Jong, J.C.; Schoufour, J.D. Macronutrient intake and frailty: The Rotterdam Study. Eur. J. Nutr. 2020, 59, 2919–2928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Yaghi, N.; Yaghi, C.; Abifadel, M.; Boulos, C.; Feart, C. Dietary Patterns and Risk Factors of Frailty in Lebanese Older Adults. Nutrients 2021, 13, 2188. [Google Scholar] [CrossRef]
  30. León-Muñoz, L.M.; García-Esquinas, E.; López-García, E.; Banegas, J.R.; Rodríguez-Artalejo, F. Major dietary patterns and risk of frailty in older adults: A prospective cohort study. BMC Med. 2015, 13, 11. [Google Scholar] [CrossRef] [Green Version]
  31. Mazidi, M.; Mikhailidis, D.P.; Sattar, N.; Toth, P.P.; Judd, S.; Blaha, M.J.; Hernandez, A.V.; Penson, P.E.; Banach, M. Association of types of dietary fats and all-cause and cause-specific mortality: A prospective cohort study and meta-analysis of prospective studies with 1,164,029 participants. Clin. Nutr. 2020, 39, 3677–3686. [Google Scholar] [CrossRef]
  32. Mozaffari, H.; Djafarian, K.; Mofrad, M.D.; Shab-Bidar, S. Dietary fat, saturated fatty acid, and monounsaturated fatty acid intakes and risk of bone fracture: A systematic review and meta-analysis of observational studies. Osteoporos Int. 2018, 29, 1949–1961. [Google Scholar] [CrossRef] [PubMed]
  33. Barnard, N.D.; Bunner, A.E.; Agarwal, U. Saturated and trans fats and dementia: A systematic review. Neurobiol. Aging 2014, 35 (Suppl. S2), S65–S73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Yuan, S.; Lemming, E.W.; Michaëlsson, K.; Larsson, S.C. Plasma phospholipid fatty acids, bone mineral density and fracture risk: Evidence from a Mendelian randomization study. Clin. Nutr. 2020, 39, 2180–2186. [Google Scholar] [CrossRef] [PubMed]
  35. Praagman, J.; Vissers, L.E.T.; Mulligan, A.A.; Laursen, A.S.D.; Beulens, J.W.J.; van der Schouw, Y.T.; Wareham, N.J.; Hansen, C.P.; Khaw, K.T.; Jakobsen, M.U.; et al. Consumption of individual saturated fatty acids and the risk of myocardial infarction in a UK and a Danish cohort. Int. J. Cardiol. 2019, 279, 18–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Aglago, E.K.; Murphy, N.; Huybrechts, I.; Nicolas, G.; Casagrande, C.; Fedirko, V.; Weiderpass, E.; Rothwell, J.A.; Dahm, C.C.; Olsen, A.; et al. Dietary intake and plasma phospholipid concentrations of saturated, monounsaturated and trans fatty acids and colorectal cancer risk in the European Prospective Investigation into Cancer and Nutrition cohort. Int. J. Cancer 2021, 149, 865–882. [Google Scholar] [CrossRef]
  37. Fretts, A.M.; Mozaffarian, D.; Siscovick, D.S.; King, I.B.; McKnight, B.; Psaty, B.M.; Rimm, E.B.; Sitlani, C.; Sacks, F.M.; Song, X.; et al. Associations of Plasma Phospholipid SFAs with Total and Cause-Specific Mortality in Older Adults Differ According to SFA Chain Length. J. Nutr. 2016, 146, 298–305. [Google Scholar] [CrossRef] [Green Version]
  38. van Rooijen, M.A.; Mensink, R.P. Palmitic Acid Versus Stearic Acid: Effects of Interesterification and Intakes on Cardiometabolic Risk Markers—A Systematic Review. Nutrients 2020, 12, 615. [Google Scholar] [CrossRef] [Green Version]
Table 1. Summary statistics of fatty acids-raising genetic variants.
Table 1. Summary statistics of fatty acids-raising genetic variants.
Effect AlleleOther AlleleFatty Acids aFrailty Index b
TraitsSNPsChrEAFβSEpβSEp
PA, 16:0rs23913881CA0.450.1780.0272.72 × 10−11−0.0110.0170.518
SA, 18:0rs66756681GT0.510.1650.0192.16 × 10−180.0160.0170.360
rs111198051TA0.880.1680.0282.80 × 10−9−0.0230.0260.371
rs10227511TC0.680.1800.0191.33 × 10−200.0700.0181.00 × 10−4
POA, 16:1n−7rs7800932TC0.410.0200.0039.80 × 10−100.0450.0180.011
rs67224562GA0.980.0480.0094.12 × 10−8−0.0280.0580.629
rs60342410GA0.810.0330.0045.69 × 10−15−0.0490.0230.033
rs709996510GA0.220.0230.0046.96 × 10−9−0.0410.0210.048
rs10227511CT0.330.0240.0036.60 × 10−13−0.0700.0181.00 × 10−4
OA, 18:1n−9rs10227511CT0.330.2300.0192.19 × 10−32−0.0700.0181.00 × 10−4
LA, 18:2n6rs1074011810GC0.560.2480.0438.08 × 10−90.0650.0172.00 × 10−4
rs17454711CT0.321.4740.0424.98 × 10−274−0.0700.0181.00 × 10−4
rs1696695216GA0.690.3510.0441.23 × 10−15−0.0230.0190.223
AA, 20:4n6rs17454711TC0.681.6910.0253.00 × 10−9710.0700.0181.00 × 10−4
rs1696695216GA0.690.1990.0312.43 × 10−10−0.0230.0190.223
ALA, 18:3n3rs17454711CT0.330.0160.0013.47 × 10−64−0.0700.0181.00 × 10−4
EPA, 20:5n3rs37987136CG0.430.0350.0051.93 × 10−120.0140.0170.434
rs17453811GA0.720.0830.0055.37 × 10−580.0660.0194.00 × 10−4
DPA, 22:5n3rs7800942TC0.410.0170.0039.04 × 10−90.0440.0180.012
rs37343986CT0.430.0400.0039.61 × 10−440.0110.0170.516
rs17454711TC0.670.0750.0033.79 × 10−1540.0700.0181.00 × 10−4
DHA, 22:6n3rs22362126GC0.570.1130.0141.26 × 10−15−0.0100.0170.571
Abbreviations: Chr = Chromosome; EAF = Effect allele frequency; SE = Standard error; SNP = Single-nucleotide polymorphism; PA = Palmitic acid; SA = Stearic acid; POA = Palmitoleic acid; OA = Oleic acid; LA = Linoleic acid; AA = Arachidonic acid; ALA = α-Linolenic acid; EPA = Eicosapentaenoic acid; DPA = Docosapentaenoic acid; DHA = Docosahexaenoic acid. a Summary statistics for PUFA from fatty acids “Circ Cardiovasc Genet 2013;6(2):171–183. doi:10.1161/CIRCGENETICS.112.964619.”, “PLoS Genet 2011;7(7):e1002193. doi:10.1371/journal.pgen.1002193” and “Circ Cardiovasc Genet 2014;7(3):321–331. doi:10.1161/CIRCGENETICS.113.000208”. b Summary statistics estimated by multiple linear regression model for frailty index from UK biobank data.
Table 2. Summary statistics for sensitivity analysis using proxy SNPs to consider the possibility of palindromic SNPs in three traits of fatty acids.
Table 2. Summary statistics for sensitivity analysis using proxy SNPs to consider the possibility of palindromic SNPs in three traits of fatty acids.
Effect AlleleOther AlleleFatty Acids aFrailty Index b
TraitsSNPsChrEAFβSEpβSEp
SA, 18:0rs66756681GT0.510.1650.0192.16 × 10−180.0160.0170.360
rs1803468 c1GA0.880.1700.0293.681 × 10−9−0.0120.0280.666
rs10227511TC0.680.1800.0191.33 × 10−200.0700.0181.00 × 10−4
LA, 18:2n6rs10761741 c10GT0.560.2510.0448.77 × 10−90.0660.0194.00 × 10−4
rs17454711CT0.321.4740.0424.98 × 10−274−0.0700.0181.00 × 10−4
rs1696695216GA0.690.3510.0441.23 × 10−15−0.0230.0190.223
EPA, 20:5n3rs4713165 c6CT0.420.0370.0064.84 × 10−110.0060.0190.753
rs17453811GA0.720.0830.0055.37 × 10−580.0660.0194.00 × 10−4
Abbreviations: Chr = Chromosome; EAF = Effect allele frequency; SE = Standard error; SNP = Single-nucleotide polymorphism; PA = Palmitic acid; SA = Stearic acid; POA = Palmitoleic acid; OA = Oleic acid; LA = Linoleic acid; AA = Arachidonic acid; ALA = α-Linolenic acid; EPA = Eicosapentaenoic acid; DPA = Docosapentaenoic acid; DHA = Docosahexaenoic acid. a Summary statistics for PUFA from fatty acids “Circ Cardiovasc Genet 2013;6(2):171–183. doi:10.1161/CIRCGENETICS.112.964619.”, “PLoS Genet 2011;7(7):e1002193. doi:10.1371/journal.pgen.1002193” and “Circ Cardiovasc Genet 2014;7(3):321–331. doi:10.1161/CIRCGENETICS.113.000208”. b Summary statistics estimated by multiple linear regression model for frailty index from UK biobank data. c Summary statistics for frailty index from UK biobank data identified by the LDproxy.
Table 3. MR results of the fatty acids and frailty index.
Table 3. MR results of the fatty acids and frailty index.
MRMultivariate MR a
β(95%CI)pβ(95%CI)p
Non-PUFAs
Saturated fatty acids
Palmitic acid (16:0)−0.063(−0.255, 0.129)0.5180.288(0.128, 0.447)<0.001
Stearic acid (18:0)0.178(0.050, 0.307)0.0070.361(0.155, 0.567)0.001
Mono-unsaturated fatty acids
Palmitoleic acid (16:1n-7)−1.127(−1.868, −0.387)0.0030.026(−1.083, 1.135)0.963
Oleic acid (18:1n-9)−0.304(−0.458, −0.150)<0.001−0.086(−0.330, 0.158)0.488
PUFAs
n-6 PUFAs
Linoleic acid (18:2n6)−0.039(−0.063, −0.016)0.0011.075(−1.549, 3.698)0.422
Arachidonic acid (20:4n6)0.039(0.018, 0.060)<0.001−0.266(−0.937, 0.406)0.438
n-3 PUFAs
α-Linolenic acid (18:3n3)−4.379(−6.615, −2.143)<0.001−44.36(−186.03, 97.32)0.539
Eicosapentaenoic acid (20:5n3)0.722(0.323, 1.122)<0.001−7.865(−50.28, 34.55)0.716
Docosapentaenoic acid (22:5n3)0.849(0.442, 1.255)<0.00123.09(−51.07, 97.25)0.542
Docosahexaenoic acid (22:6n3)−0.088(−0.390, 0.215)0.5714.482(−5.569, 14.533)0.382
Abbreviations: MR = Mendelian randomization with inverse variance weighted method using a fixed-effect model; 95% CI = 95% confidence interval; Multivariate MR = Multivariable Mendelian randomization. PUFA = polyunsaturated fatty acids. a Multivariate model for non-PUFAs included palmitic acid, stearic acid, palmitoleic acid, and oleic acid as exposure variables. Multivariate model for PUFAs included linoleic acid, arachidonic acid, α-Linolenic acid, eicosapentaenoic acid, docosapentaenoic acid, and docosahexaenoic acid as exposure variables.
Table 4. Sensitivity analysis: MR results of the fatty acids and frailty index using the proxy SNPs.
Table 4. Sensitivity analysis: MR results of the fatty acids and frailty index using the proxy SNPs.
MRMultivariate MR a
β(95%CI)pβ(95%CI)p
Stearic acid (18:0)0.177(0.049, 0.305)0.0070.360(0.154, 0.566)0.001
Linoleic acid (18:2n6)−0.039(−0.062, −0.016)0.001−0.046(−0.927, 0.835)0.919
Eicosapentaenoic acid (20:5n3)0.658(0.263, 1.054)0.0019.832(−3.754, 23.417)0.156
Abbreviations: MR = Mendelian randomization with inverse variance weighted method using a fixed-effect model; 95% CI = 95% confidence interval; Multivariate MR = Multivariable Mendelian randomization. PUFA = polyunsaturated fatty acids. a Models were the same as the multivariate MR in Table 3.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tomata, Y.; Wang, Y.; Hägg, S.; Jylhävä, J. Fatty Acids and Frailty: A Mendelian Randomization Study. Nutrients 2021, 13, 3539. https://doi.org/10.3390/nu13103539

AMA Style

Tomata Y, Wang Y, Hägg S, Jylhävä J. Fatty Acids and Frailty: A Mendelian Randomization Study. Nutrients. 2021; 13(10):3539. https://doi.org/10.3390/nu13103539

Chicago/Turabian Style

Tomata, Yasutake, Yunzhang Wang, Sara Hägg, and Juulia Jylhävä. 2021. "Fatty Acids and Frailty: A Mendelian Randomization Study" Nutrients 13, no. 10: 3539. https://doi.org/10.3390/nu13103539

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop